1
|
Sultan M, Razzaq M, Lee J, Das S, Kannappan S, Subramani VK, Yoo W, Kim T, Lee HR, Chaurasia AK, Kim KK. Targeting the G-quadruplex as a novel strategy for developing antibiotics against hypervirulent drug-resistant Staphylococcus aureus. J Biomed Sci 2025; 32:15. [PMID: 39905515 DOI: 10.1186/s12929-024-01109-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 12/09/2024] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND The rapid emergence of multiple drug-resistant (MDR) bacterial pathogens and the lack of a novel antibiotic pipeline pose a serious threat to global healthcare. The limited number of established targets further restricts the identification of novel antibiotics to treat life-threatening MDR infections caused by Staphylococcus aureus strains. Therefore, novel targets for developing antibiotics are urgently required. In this study, we hypothesized that the G-quadruplex (G4)-binding ligands can be used as novel antibiotics as their binding can possibly downregulate/block the expression of vital genes. METHODS To test this, first we screened the antibiotic properties of representative G4-binding ligands against hypervirulent and MDR S. aureus USA300 and determined the in vitro and in vivo antibacterial activity; and proposed the mechanism of action by applying various microbiological, infection, microscopic, and biophysicochemical techniques. RESULTS Herein, among screened G4-binding ligands, N-methyl mesoporphyrin IX (NMM) showed the highest antibacterial activity against S. aureus USA300. NMM exhibited a minimum inhibitory concentration (MIC) of 5 μM against S. aureus USA300, impacting cell division and the cell wall by repressing the expressions of genes in the division cell wall (dcw) gene cluster. Genome-wide bioinformatics analysis of G4 motifs and their mapping on S. aureus genome, identified the presence of G4-motif in the promoter of mraZ, a conserved master regulator of the dcw cluster regulating the coordinated cell division and cell wall synthesis. Physicochemical assessments using UV-visible, circular dichroism, and nuclear magnetic resonance spectroscopy confirmed that the G4-motif present in the mraZ promoter formed an intramolecular parallel G4 structure, interacting with NMM. In vivo reporter followed by coupled in vitro transcription/translation (IVT) assays confirmed the role of mraZ G4 as a target interacting NMM to impose extreme antibacterial activity against both the gram-positive and -negative bacteria. In-cell and in vivo validation of NMM using RAW264.7 cells and Galleria mellonella; respectively, demonstrated that NMM exhibited superior antibiotic activity compared to well-established antibiotics, with no observed cytotoxicity. CONCLUSIONS In summary, the current study identified NMM as a broad-spectrum potent antibacterial agent and elucidated its plausible mechanism of action primarily by targeting G4-motif in the mraZ promoter of the dcw gene cluster.
Collapse
Affiliation(s)
- Maria Sultan
- Department of Precision Medicine, Graduate School of Basic Medical Science, Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Maria Razzaq
- Department of Precision Medicine, Graduate School of Basic Medical Science, Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Joohyun Lee
- Department of Precision Medicine, Graduate School of Basic Medical Science, Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Shreyasi Das
- Department of Precision Medicine, Graduate School of Basic Medical Science, Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Shrute Kannappan
- Department of Precision Medicine, Graduate School of Basic Medical Science, Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Vinod Kumar Subramani
- Department of Precision Medicine, Graduate School of Basic Medical Science, Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Wanki Yoo
- Department of Precision Medicine, Graduate School of Basic Medical Science, Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Truc Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science, Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Hye-Ra Lee
- Department of Biotechnology and Bioinformatics, College of Science and Technology, Korea University, Sejong, 30019, Republic of Korea
| | - Akhilesh K Chaurasia
- Department of Precision Medicine, Graduate School of Basic Medical Science, Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science, Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
| |
Collapse
|
2
|
Wang YT, Liu LT, Hou B, Yao CM, Wang XF, Lu B. Recent advances in studies on FtsZ inhibitors. Biochem Pharmacol 2024; 230:116551. [PMID: 39307317 DOI: 10.1016/j.bcp.2024.116551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 10/02/2024]
Abstract
With the abuse of antibiotics, multidrug resistant strains continue to emerge and spread rapidly. Therefore, there is an urgent need to develop new antimicrobial drugs. As a highly conserved cell division protein in bacteria, filamenting temperature-sensitive mutant Z (FtsZ) has been identified as a potential antimicrobial target. This paper reviews the structure, function, and action mechanism of FtsZ and a variety of natural and synthetic compounds targeting FtsZ, including 3-MBA derivatives, taxane derivatives, cinnamaldehyde, curcumin, quinoline and quinazoline derivatives, aromatic compounds, purpurin, and totarol. From these studies, FtsZ has a clear supporting role in the field of antimicrobial drug discovery. The urgent need and interest of antibacterial drugs will contribute to the discovery of new clinical drugs targeting FtsZ.
Collapse
Affiliation(s)
- Yan-Ting Wang
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai 200433, PR China.
| | - Lan-Tian Liu
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai 200433, PR China
| | - Bo Hou
- School of Life Science and Technology, Xidian University, Xi'an 710126, PR China
| | - Chun-Meng Yao
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai 200433, PR China
| | - Xu-Fang Wang
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai 200433, PR China
| | - Bin Lu
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai 200433, PR China.
| |
Collapse
|
3
|
Basak P, Dastidar DG, Ghosh D, Chakraborty T, Sau S, Chakrabarti G. Staphylococcus aureus major cell division protein FtsZ assembly is inhibited by silibinin, a natural flavonolignan that also blocked bacterial growth and biofilm formation. Int J Biol Macromol 2024; 279:135252. [PMID: 39222779 DOI: 10.1016/j.ijbiomac.2024.135252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
The bacterial cell division protein FtsZ has been considered a potential therapeutic target due to its rapid treadmilling that induces cellular wall construction in bacteria. The current study discovered a novel antimicrobial compound, silibinin, a natural flavonolignan and its impact on the recombinant S. aureus FtsZ (SaFtsZ). Silibinin inhibited S. aureus Newman growth in a dose-dependent manner. The IC50 and MIC values for silibinin were 75 μM and 200 μM, respectively. It had no cytotoxicity against HEK293 cells in vitro. Silibinin also enlarged the bacterial cell morphology by ∼40 folds and showed antibiofilm property. It perturbed the S. aureus membrane potential both at IC50 conc. and at MIC conc. Further, it inhibited both the polymerization and GTPase activity of SaFtsZ. It did not inhibit tubulin assembly, a eukaryotic FtsZ homolog. A fluorescence quenching study yielded the Kd value for SaFtsZ-Silibinin interaction and binding stoichiometry 0.857 ± 0.188 μM and 1:1, respectively. Both in silico study and competition assay indicated that silibinin binds at the GTP binding site on SaFtsZ. The Ki value for the silibinin-mediated inhibition of SaFtsZ was 8.8 μM. Therefore, these findings have comprehensively shown the antimicrobial behavior of silibinin on S. aureus Newman cells targeting SaFtsZ.
Collapse
Affiliation(s)
- Prithvi Basak
- Department of Biotechnology, Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, WB 700 019, India
| | - Debabrata Ghosh Dastidar
- Guru Nanak Institute of Pharmaceutical Science & Technology, 157/F Nilgunj Road, Panihati, Kolkata 700114, West Bengal, India
| | - Dipanjan Ghosh
- Department of Biotechnology, Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, WB 700 019, India
| | - Tushar Chakraborty
- Department of Biological Sciences, Bose Institute, Kolkata 700091, West Bengal, India
| | - Subrata Sau
- Department of Biological Sciences, Bose Institute, Kolkata 700091, West Bengal, India
| | - Gopal Chakrabarti
- Department of Biotechnology, Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, WB 700 019, India.
| |
Collapse
|
4
|
Shinde Y, Pathan A, Chinnam S, Rathod G, Patil B, Dhangar M, Mathew B, Kim H, Mundada A, Kukreti N, Ahmad I, Patel H. Mycobacterial FtsZ and inhibitors: a promising target for the anti-tubercular drug development. Mol Divers 2024; 28:3457-3478. [PMID: 38010605 DOI: 10.1007/s11030-023-10759-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/30/2023] [Indexed: 11/29/2023]
Abstract
The emergence of multidrug-resistant tuberculosis (MDR-TB) strains has rendered many anti-TB drugs ineffective. Consequently, there is an urgent need to identify new drug targets against Mycobacterium tuberculosis (Mtb). Filament Forming Temperature Sensitive Gene Z (FtsZ), a member of the cytoskeletal protein family, plays a vital role in cell division by forming a cytokinetic ring at the cell's center and coordinating the division machinery. When FtsZ is depleted, cells are unable to divide and instead elongate into filamentous structures that eventually undergo lysis. Since the inactivation of FtsZ or alterations in its assembly impede the formation of the Z-ring and septum, FtsZ shows promise as a target for the development of anti-mycobacterial drugs. This review not only discusses the potential role of FtsZ as a promising pharmacological target for anti-tuberculosis therapies but also explores the structural and functional aspects of the mycobacterial protein FtsZ in cell division. Additionally, it reviews various inhibitors of Mtb FtsZ. By understanding the importance of FtsZ in cell division, researchers can explore strategies to disrupt its function, impeding the growth and proliferation of Mtb. Furthermore, the investigation of different inhibitors that target Mtb FtsZ expands the potential for developing effective treatments against tuberculosis.
Collapse
Affiliation(s)
- Yashodeep Shinde
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, 425405, India
| | - Asama Pathan
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, 425405, India
| | - Sampath Chinnam
- Department of Chemistry, M. S. Ramaiah Institute of Technology (Autonomous Institute, Affiliated to Visvesvaraya Technological University, Belgaum), Bengaluru, Karnataka, 560054, India
| | - Gajanan Rathod
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Mohali, Punjab, 160062, India
| | - Bhatu Patil
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, 425405, India
| | - Mayur Dhangar
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, 425405, India
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 690525, India
| | - Hoon Kim
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon, 57922, Republic of Korea
| | - Anand Mundada
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, 425405, India
| | - Neelima Kukreti
- School of Pharmacy, Graphic Era Hill University-Dehradun, Dehradun, Uttarakhand, 248002, India
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, 425405, India
| | - Harun Patel
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, 425405, India.
| |
Collapse
|
5
|
Meng J, Li M, Zheng Z, Sun Z, Yang S, Ouyang G, Wang Z, Zhou X. Application of natural-products repurposing strategy to discover novel FtsZ inhibitors: Bactericidal evaluation and the structure-activity relationship of sanguinarine and its analogs. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 203:106016. [PMID: 39084807 DOI: 10.1016/j.pestbp.2024.106016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/21/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024]
Abstract
The novel bactericidal target-filamentous temperature-sensitive protein Z (FtsZ)-has drawn the attention of pharmacologists to address the emerging issues with drug/pesticide resistance caused by pathogenic bacteria. To enrich the structural diversity of FtsZ inhibitors, the antibacterial activity and structure-activity relationship (SAR) of natural sanguinarine and its analogs were investigated by using natural-products repurposing strategy. Notably, sanguinarine and chelerythrine exerted potent anti-Xanthomonas oryzae pv. oryzae (Xoo) activity, with EC50 values of 0.96 and 0.93 mg L-1, respectively, among these molecules. Furthermore, these two compounds could inhibit the GTPase activity of XooFtsZ, with IC50 values of 241.49 μM and 283.14 μM, respectively. An array of bioassays including transmission electron microscopy (TEM), fluorescence titration, and Fourier transform infrared spectroscopy (FT-IR) co-verified that sanguinarine and chelerythrine were potential XooFtsZ inhibitors that could interfere with the assembly of FtsZ filaments by inhibiting the GTPase hydrolytic ability of XooFtsZ protein. Additionally, the pot experiment suggested that chelerythrine and sanguinarine demonstrated excellent curative activity with values of 59.52% and 54.76%, respectively. Excitedly, these two natural compounds also showed outstanding druggability, validated by acceptable drug-like properties and low toxicity on rice. Overall, the results suggested that chelerythrine was a new and potential XooFtsZ inhibitor to develop new bactericide and provided important guiding values for rational drug design of FtsZ inhibitors. Notably, our findings provide a novel strategy to discover novel, promising and green bacterial compounds for the management of plant bacterial diseases.
Collapse
Affiliation(s)
- Jiao Meng
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Mei Li
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Zhicheng Zheng
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Zhaoju Sun
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Song Yang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Guiping Ouyang
- School of Pharmaceutical Sciences, Guizhou University, Huaxi District, Guiyang, 550025, China.
| | - Zhenchao Wang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China; School of Pharmaceutical Sciences, Guizhou University, Huaxi District, Guiyang, 550025, China.
| | - Xiang Zhou
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China.
| |
Collapse
|
6
|
Weber F, Weber A, Schmitt L, Lechtenberg I, Greb J, Henßen B, Wesselborg S, Pietruszka J. From the Total Synthesis of Semi-Viriditoxin, Semi-Viriditoxic Acid and Dimeric Naphthopyranones to their Biological Activities in Burkitt B Cell Lymphoma. Chemistry 2024; 30:e202400559. [PMID: 38411573 DOI: 10.1002/chem.202400559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 02/28/2024]
Abstract
Dimeric naphthopyranones are known to be biologically active, however, for the corresponding monomeric naphthopyranones this information is still elusive. Here the first enantioselective total synthesis of semi-viriditoxic acid as well as the synthesis of semi-viriditoxin and derivatives is reported. The key intermediate in the synthesis of naphthopyranones is an α,β-unsaturated δ-lactone, which we synthesized in two different ways (Ghosez-cyclization and Grubbs ring-closing metathesis), while the domino-Michael-Dieckmann reaction of the α,β-unsaturated δ-lactone with an orsellinic acid derivative is the key reaction. A structure-activity relationship study was performed measuring the cytotoxicity in Burkitt B lymphoma cells (Ramos). The dimeric structure was found to be crucial for biological activity: Only the dimeric naphthopyranones showed cytotoxic and apoptotic activity, whereas the monomers did not display any activity at all.
Collapse
Affiliation(s)
- Frederike Weber
- Institute for Bioorganic Chemistry, Heinrich Heine University Düsseldorf in, Forschungszentrum Jülich, 52426, Jülich, Germany
| | - Anja Weber
- Institute for Bioorganic Chemistry, Heinrich Heine University Düsseldorf in, Forschungszentrum Jülich, 52426, Jülich, Germany
| | - Laura Schmitt
- Institute for Molecular Medicine I, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Ilka Lechtenberg
- Institute for Molecular Medicine I, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Julian Greb
- Institute for Bioorganic Chemistry, Heinrich Heine University Düsseldorf in, Forschungszentrum Jülich, 52426, Jülich, Germany
| | - Birgit Henßen
- Institute for Bioorganic Chemistry, Heinrich Heine University Düsseldorf in, Forschungszentrum Jülich, 52426, Jülich, Germany
- Institute of Bio- and Geosciences (IBG-1: Biotechnology), Forschungszentrum Jülich, 52426, Jülich, Germany
| | - Sebastian Wesselborg
- Institute for Molecular Medicine I, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Jörg Pietruszka
- Institute for Bioorganic Chemistry, Heinrich Heine University Düsseldorf in, Forschungszentrum Jülich, 52426, Jülich, Germany
- Institute of Bio- and Geosciences (IBG-1: Biotechnology), Forschungszentrum Jülich, 52426, Jülich, Germany
| |
Collapse
|
7
|
Carson MC, Kozlowski MC. Recent advances in oxidative phenol coupling for the total synthesis of natural products. Nat Prod Rep 2024; 41:208-227. [PMID: 37294301 PMCID: PMC10709532 DOI: 10.1039/d3np00009e] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Covering: 2008 to 2023This review will describe oxidative phenol coupling as applied in the total synthesis of natural products. This review covers catalytic and electrochemical methods with a brief comparison to stoichiometric and enzymatic systems assessing their practicality, atom economy, and other measures. Natural products forged by C-C and C-O oxidative phenol couplings as well as from alkenyl phenol couplings will be addressed. Additionally, exploration into catalytic oxidative coupling of phenols and other related species (carbazoles, indoles, aryl ethers, etc.) will be surveyed. Future directions of this particular area of research will also be assessed.
Collapse
Affiliation(s)
- Matthew C Carson
- Department of Chemistry, Roy and Diana Vagelos Laboratories, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, USA.
| | - Marisa C Kozlowski
- Department of Chemistry, Roy and Diana Vagelos Laboratories, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, USA.
| |
Collapse
|
8
|
Wennrich JP, Sepanian E, Ebada SS, Llanos-Lopez NA, Ashrafi S, Maier W, Kurtán T, Stadler M. Bioactive Naphtho-α-Pyranones from Two Endophytic Fungi of the Genus Polyphilus. Antibiotics (Basel) 2023; 12:1273. [PMID: 37627693 PMCID: PMC10451773 DOI: 10.3390/antibiotics12081273] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
In the course of our survey to study the metabolic potential of two species of a new helotialean genus Polyphilus, namely P. frankenii and P. sieberi, their crude extracts were obtained using different cultivation techniques, which led to the isolation and characterization of two new naphtho-α-pyranone derivatives recognized as a monomer (1) and its 6,6'-homodimer (2) together with two known diketopiperazine congeners, outovirin B (3) and (3S,6S)-3,6-dibenzylpiperazine-2,5-dione (4). The structures of isolated compounds were determined based on extensive 1D and 2D NMR and HRESIMS. The absolute configuration of new naphtho-α-pyranones was determined using a comparison of their experimental ECD spectra with those of related structural analogues. 6,6'-binaphtho-α-pyranone talaroderxine C (2) exhibited potent cytotoxic activity against different mammalian cell lines with IC50 values in the low micromolar to nanomolar range. In addition, talaroderxine C unveiled stronger antimicrobial activity against Bacillus subtilis rather than Staphylococcus aureus with MIC values of 0.52 µg mL-1 (0.83 µM) compared to 66.6 µg mL-1 (105.70 µM), respectively.
Collapse
Affiliation(s)
- Jan-Peer Wennrich
- Department of Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Inhoffenstrasse 7, 38124 Braunschweig, Germany; (J.-P.W.); (E.S.); (N.A.L.-L.)
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| | - Ellen Sepanian
- Department of Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Inhoffenstrasse 7, 38124 Braunschweig, Germany; (J.-P.W.); (E.S.); (N.A.L.-L.)
| | - Sherif S. Ebada
- Department of Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Inhoffenstrasse 7, 38124 Braunschweig, Germany; (J.-P.W.); (E.S.); (N.A.L.-L.)
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Natalia A. Llanos-Lopez
- Department of Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Inhoffenstrasse 7, 38124 Braunschweig, Germany; (J.-P.W.); (E.S.); (N.A.L.-L.)
| | - Samad Ashrafi
- Institute for Epidemiology and Pathogen Diagnostics, Julius Kühn Institute (JKI)—Federal Research Centre for Cultivated Plants, Messeweg 11-12, 38104 Braunschweig, Germany; (S.A.); (W.M.)
- Institute for Crop and Soil Science, Julius Kühn Institute (JKI)—Federal Research Centre for Cultivated Plants, Bundesallee 58, 38116 Braunschweig, Germany
| | - Wolfgang Maier
- Institute for Epidemiology and Pathogen Diagnostics, Julius Kühn Institute (JKI)—Federal Research Centre for Cultivated Plants, Messeweg 11-12, 38104 Braunschweig, Germany; (S.A.); (W.M.)
| | - Tibor Kurtán
- Department of Organic Chemistry, University of Debrecen, P.O. Box 400, 4002 Debrecen, Hungary;
| | - Marc Stadler
- Department of Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Inhoffenstrasse 7, 38124 Braunschweig, Germany; (J.-P.W.); (E.S.); (N.A.L.-L.)
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| |
Collapse
|
9
|
Kifayat S, Yele V, Ashames A, Sigalapalli DK, Bhandare RR, Shaik AB, Nasipireddy V, Sanapalli BKR. Filamentous temperature sensitive mutant Z: a putative target to combat antibacterial resistance. RSC Adv 2023; 13:11368-11384. [PMID: 37057268 PMCID: PMC10089256 DOI: 10.1039/d3ra00013c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/28/2023] [Indexed: 04/15/2023] Open
Abstract
In the pre-antibiotic era, common bacterial infections accounted for high mortality and morbidity. Moreover, the discovery of penicillin in 1928 marked the beginning of an antibiotic revolution, and this antibiotic era witnessed the discovery of many novel antibiotics, a golden era. However, the misuse or overuse of these antibiotics, natural resistance that existed even before the antibiotics were discovered, genetic variations in bacteria, natural selection, and acquisition of resistance from one species to another consistently increased the resistance to the existing antibacterial targets. Antibacterial resistance (ABR) is now becoming an ever-increasing concern jeopardizing global health. Henceforth, there is an urgent unmet need to discover novel compounds to combat ABR, which act through untapped pathways/mechanisms. Filamentous Temperature Sensitive mutant Z (FtsZ) is one such unique target, a tubulin homolog involved in developing a cytoskeletal framework for the cytokinetic ring. Additionally, its pivotal role in bacterial cell division and the lack of homologous structural protein in mammals makes it a potential antibacterial target for developing novel molecules. Approximately 2176 X-crystal structures of FtsZ were available, which initiated the research efforts to develop novel antibacterial agents. The literature has reported several natural, semisynthetic, peptides, and synthetic molecules as FtsZ inhibitors. This review provides valuable insights into the basic crystal structure of FtsZ, its inhibitors, and their inhibitory activities. This review also describes the available in vitro detection and quantification methods of FtsZ-drug complexes and the various approaches for determining drugs targeting FtsZ polymerization.
Collapse
Affiliation(s)
- Sumaiya Kifayat
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University Rajasthan Jaipur 303121 India +91-9291661992
| | - Vidyasrilekha Yele
- Department of Pharmaceutical Chemistry, NIMS Institute of Pharmacy, NIMS University Rajasthan Jaipur 303121 India
| | - Akram Ashames
- College of Pharmacy & Health Sciences, Ajman University PO Box 340 Ajman United Arab Emirates
- Center of Medical and Bio-allied Health Sciences Research, Ajman University PO Box 340 Ajman United Arab Emirates +97167056240
| | - Dilep Kumar Sigalapalli
- Department of Pharmaceutical Chemistry, Vignan Pharmacy College, Jawaharlal Nehru Technological University Vadlamudi 522213 Andhra Pradesh India
| | - Richie R Bhandare
- College of Pharmacy & Health Sciences, Ajman University PO Box 340 Ajman United Arab Emirates
- Center of Medical and Bio-allied Health Sciences Research, Ajman University PO Box 340 Ajman United Arab Emirates +97167056240
| | - Afzal B Shaik
- St. Mary's College of Pharmacy, St. Mary's Group of Institutions Guntur, Affiliated to Jawaharlal Nehru Technological University Kakinada Chebrolu Guntur 522212 Andhra Pradesh India
| | | | - Bharat Kumar Reddy Sanapalli
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University Rajasthan Jaipur 303121 India +91-9291661992
| |
Collapse
|
10
|
Sharma AK, Poddar SM, Chakraborty J, Nayak BS, Kalathil S, Mitra N, Gayathri P, Srinivasan R. A mechanism of salt bridge-mediated resistance to FtsZ inhibitor PC190723 revealed by a cell-based screen. Mol Biol Cell 2023; 34:ar16. [PMID: 36652338 PMCID: PMC10011733 DOI: 10.1091/mbc.e22-12-0538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Bacterial cell division proteins, especially the tubulin homologue FtsZ, have emerged as strong targets for developing new antibiotics. Here, we have utilized the fission yeast heterologous expression system to develop a cell-based assay to screen for small molecules that directly and specifically target the bacterial cell division protein FtsZ. The strategy also allows for simultaneous assessment of the toxicity of the drugs to eukaryotic yeast cells. As a proof-of-concept of the utility of this assay, we demonstrate the effect of the inhibitors sanguinarine, berberine, and PC190723 on FtsZ. Though sanguinarine and berberine affect FtsZ polymerization, they exert a toxic effect on the cells. Further, using this assay system, we show that PC190723 affects Helicobacter pylori FtsZ function and gain new insights into the molecular determinants of resistance to PC190723. On the basis of sequence and structural analysis and site-specific mutations, we demonstrate that the presence of salt bridge interactions between the central H7 helix and β-strands S9 and S10 mediates resistance to PC190723 in FtsZ. The single-step in vivo cell-based assay using fission yeast enabled us to dissect the contribution of sequence-specific features of FtsZ and cell permeability effects associated with bacterial cell envelopes. Thus, our assay serves as a potent tool to rapidly identify novel compounds targeting polymeric bacterial cytoskeletal proteins like FtsZ to understand how they alter polymerization dynamics and address resistance determinants in targets.
Collapse
Affiliation(s)
- Ajay Kumar Sharma
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Centre for Interdisciplinary Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Homi Bhabha National Institutes, Anushakti Nagar, Mumbai 400094, India
| | - Sakshi Mahesh Poddar
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Centre for Interdisciplinary Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Homi Bhabha National Institutes, Anushakti Nagar, Mumbai 400094, India
| | - Joyeeta Chakraborty
- Biology, Indian Institute of Science Education and Research, Pune 411008, India
| | - Bhagyashri Soumya Nayak
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Centre for Interdisciplinary Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Homi Bhabha National Institutes, Anushakti Nagar, Mumbai 400094, India
| | - Srilakshmi Kalathil
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Centre for Interdisciplinary Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Homi Bhabha National Institutes, Anushakti Nagar, Mumbai 400094, India
| | - Nivedita Mitra
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Centre for Interdisciplinary Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Homi Bhabha National Institutes, Anushakti Nagar, Mumbai 400094, India
| | - Pananghat Gayathri
- Biology, Indian Institute of Science Education and Research, Pune 411008, India
| | - Ramanujam Srinivasan
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Centre for Interdisciplinary Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Homi Bhabha National Institutes, Anushakti Nagar, Mumbai 400094, India
| |
Collapse
|
11
|
Models versus pathogens: how conserved is the FtsZ in bacteria? Biosci Rep 2023; 43:232502. [PMID: 36695643 PMCID: PMC9939409 DOI: 10.1042/bsr20221664] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/10/2023] [Accepted: 01/25/2023] [Indexed: 01/26/2023] Open
Abstract
Combating anti-microbial resistance by developing alternative strategies is the need of the hour. Cell division, particularly FtsZ, is being extensively studied for its potential as an alternative target for anti-bacterial therapy. Bacillus subtilis and Escherichia coli are the two well-studied models for research on FtsZ, the leader protein of the cell division machinery. As representatives of gram-positive and gram-negative bacteria, respectively, these organisms have provided an extensive outlook into the process of cell division in rod-shaped bacteria. However, research on other shapes of bacteria, like cocci and ovococci, lags behind that of model rods. Even though most regions of FtsZ show sequence and structural conservation throughout bacteria, the differences in FtsZ functioning and interacting partners establish several different modes of division in different bacteria. In this review, we compare the features of FtsZ and cell division in the model rods B. subtilis and E. coli and the four pathogens: Staphylococcus aureus, Streptococcus pneumoniae, Mycobacterium tuberculosis, and Pseudomonas aeruginosa. Reviewing several recent articles on these pathogenic bacteria, we have highlighted the functioning of FtsZ, the unique roles of FtsZ-associated proteins, and the cell division processes in them. Further, we provide a detailed look at the anti-FtsZ compounds discovered and their target bacteria, emphasizing the need for elucidation of the anti-FtsZ mechanism of action in different bacteria. Current challenges and opportunities in the ongoing journey of identifying potent anti-FtsZ drugs have also been described.
Collapse
|
12
|
Wang Z, Meng L, Liu X, Zhang L, Yu Z, Wu G. Recent progress toward developing axial chirality bioactive compounds. Eur J Med Chem 2022; 243:114700. [DOI: 10.1016/j.ejmech.2022.114700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 11/03/2022]
|
13
|
The mycotoxin viriditoxin induces leukemia- and lymphoma-specific apoptosis by targeting mitochondrial metabolism. Cell Death Dis 2022; 13:938. [PMID: 36347842 PMCID: PMC9643474 DOI: 10.1038/s41419-022-05356-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 09/30/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022]
Abstract
Inhibition of the mitochondrial metabolism offers a promising therapeutic approach for the treatment of cancer. Here, we identify the mycotoxin viriditoxin (VDT), derived from the endophytic fungus Cladosporium cladosporioides, as an interesting candidate for leukemia and lymphoma treatment. VDT displayed a high cytotoxic potential and rapid kinetics of caspase activation in Jurkat leukemia and Ramos lymphoma cells in contrast to solid tumor cells that were affected to a much lesser extent. Most remarkably, human hematopoietic stem and progenitor cells and peripheral blood mononuclear cells derived from healthy donors were profoundly resilient to VDT-induced cytotoxicity. Likewise, the colony-forming capacity was affected only at very high concentrations, which provides a therapeutic window for cancer treatment. Intriguingly, VDT could directly activate the mitochondrial apoptosis pathway in leukemia cells in the presence of antiapoptotic Bcl-2 proteins. The mitochondrial toxicity of VDT was further confirmed by inhibition of mitochondrial respiration, breakdown of the mitochondrial membrane potential (ΔΨm), the release of mitochondrial cytochrome c, generation of reactive oxygen species (ROS), processing of the dynamin-like GTPase OPA1 and subsequent fission of mitochondria. Thus, VDT-mediated targeting of mitochondrial oxidative phosphorylation (OXPHOS) might represent a promising therapeutic approach for the treatment of leukemia and lymphoma without affecting hematopoietic stem and progenitor cells.
Collapse
|
14
|
Chiang YM, Lin TS, Wang CCC. Total Heterologous Biosynthesis of Fungal Natural Products in Aspergillus nidulans. JOURNAL OF NATURAL PRODUCTS 2022; 85:2484-2518. [PMID: 36173392 PMCID: PMC9621686 DOI: 10.1021/acs.jnatprod.2c00487] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Fungal natural products comprise a wide range of bioactive compounds including important drugs and agrochemicals. Intriguingly, bioinformatic analyses of fungal genomes have revealed that fungi have the potential to produce significantly more natural products than what have been discovered so far. It has thus become widely accepted that most biosynthesis pathways of fungal natural products are silent or expressed at very low levels under laboratory cultivation conditions. To tap into this vast chemical reservoir, the reconstitution of entire biosynthetic pathways in genetically tractable fungal hosts (total heterologous biosynthesis) has become increasingly employed in recent years. This review summarizes total heterologous biosynthesis of fungal natural products accomplished before 2020 using Aspergillus nidulans as heterologous hosts. We review here Aspergillus transformation, A. nidulans hosts, shuttle vectors for episomal expression, and chromosomal integration expression. These tools, collectively, not only facilitate the discovery of cryptic natural products but can also be used to generate high-yield strains with clean metabolite backgrounds. In comparison with total synthesis, total heterologous biosynthesis offers a simplified strategy to construct complex molecules and holds potential for commercial application.
Collapse
Affiliation(s)
- Yi-Ming Chiang
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089, United States
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan
| | - Tzu-Shyang Lin
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089, United States
| | - Clay C C Wang
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089, United States
- Department of Chemistry, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, California 90089, United States
| |
Collapse
|
15
|
Andreu JM, Huecas S, Araújo-Bazán L, Vázquez-Villa H, Martín-Fontecha M. The Search for Antibacterial Inhibitors Targeting Cell Division Protein FtsZ at Its Nucleotide and Allosteric Binding Sites. Biomedicines 2022; 10:1825. [PMID: 36009372 PMCID: PMC9405007 DOI: 10.3390/biomedicines10081825] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022] Open
Abstract
The global spread of bacterial antimicrobial resistance is associated to millions of deaths from bacterial infections per year, many of which were previously treatable. This, combined with slow antibiotic deployment, has created an urgent need for developing new antibiotics. A still clinically unexploited mode of action consists in suppressing bacterial cell division. FtsZ, an assembling GTPase, is the key protein organizing division in most bacteria and an attractive target for antibiotic discovery. Nevertheless, developing effective antibacterial inhibitors targeting FtsZ has proven challenging. Here we review our decade-long multidisciplinary research on small molecule inhibitors of bacterial division, in the context of global efforts to discover FtsZ-targeting antibiotics. We focus on methods to characterize synthetic inhibitors that either replace bound GTP from the FtsZ nucleotide binding pocket conserved across diverse bacteria or selectively bind into the allosteric site at the interdomain cleft of FtsZ from Bacillus subtilis and the pathogen Staphylococcus aureus. These approaches include phenotype screening combined with fluorescence polarization screens for ligands binding into each site, followed by detailed cytological profiling, and biochemical and structural studies. The results are analyzed to design an optimized workflow to identify effective FtsZ inhibitors, and new approaches for the discovery of FtsZ-targeting antibiotics are discussed.
Collapse
Affiliation(s)
- José M. Andreu
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040 Madrid, Spain; (S.H.); (L.A.-B.)
| | - Sonia Huecas
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040 Madrid, Spain; (S.H.); (L.A.-B.)
| | - Lidia Araújo-Bazán
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040 Madrid, Spain; (S.H.); (L.A.-B.)
| | - Henar Vázquez-Villa
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040 Madrid, Spain;
| | - Mar Martín-Fontecha
- Departamento de Química Orgánica, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040 Madrid, Spain
| |
Collapse
|
16
|
Juskewitz E, Mishchenko E, Dubey VK, Jenssen M, Jakubec M, Rainsford P, Isaksson J, Andersen JH, Ericson JU. Lulworthinone: In Vitro Mode of Action Investigation of an Antibacterial Dimeric Naphthopyrone Isolated from a Marine Fungus. Mar Drugs 2022; 20:md20050277. [PMID: 35621928 PMCID: PMC9147123 DOI: 10.3390/md20050277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 01/27/2023] Open
Abstract
Treatment options for infections caused by antimicrobial-resistant bacteria are rendered ineffective, and drug alternatives are needed—either from new chemical classes or drugs with new modes of action. Historically, natural products have been important contributors to drug discovery. In a recent study, the dimeric naphthopyrone lulworthinone produced by an obligate marine fungus in the family Lulworthiaceae was discovered. The observed potent antibacterial activity against Gram-positive bacteria, including several clinical methicillin-resistant Staphylococcus aureus (MRSA) isolates, prompted this follow-up mode of action investigation. This paper aimed to characterize the antibacterial mode of action (MOA) of lulworthinone by combining in vitro assays, NMR experiments and microscopy. The results point to a MOA targeting the bacterial membrane, leading to improper cell division. Treatment with lulworthinone induced an upregulation of genes responding to cell envelope stress in Bacillus subtilis. Analysis of the membrane integrity and membrane potential indicated that lulworthinone targets the bacterial membrane without destroying it. This was supported by NMR experiments using artificial lipid bilayers. Fluorescence microscopy revealed that lulworthinone affects cell morphology and impedes the localization of the cell division protein FtsZ. Surface plasmon resonance and dynamic light scattering assays showed that this activity is linked with the compound‘s ability to form colloidal aggregates. Antibacterial agents acting at cell membranes are of special interest, as the development of bacterial resistance to such compounds is deemed more difficult to occur.
Collapse
Affiliation(s)
- Eric Juskewitz
- Research Group for Host Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, 9019 Tromsø, Norway; (E.M.); (V.K.D.)
- Correspondence: (E.J.); (J.U.E.)
| | - Ekaterina Mishchenko
- Research Group for Host Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, 9019 Tromsø, Norway; (E.M.); (V.K.D.)
| | - Vishesh K. Dubey
- Research Group for Host Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, 9019 Tromsø, Norway; (E.M.); (V.K.D.)
| | - Marte Jenssen
- Marbio, The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT the Arctic University of Norway, 9019 Tromsø, Norway; (M.J.); (J.H.A.)
| | - Martin Jakubec
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, 9019 Tromsø, Norway; (M.J.); (P.R.); (J.I.)
| | - Philip Rainsford
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, 9019 Tromsø, Norway; (M.J.); (P.R.); (J.I.)
| | - Johan Isaksson
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, 9019 Tromsø, Norway; (M.J.); (P.R.); (J.I.)
| | - Jeanette H. Andersen
- Marbio, The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT the Arctic University of Norway, 9019 Tromsø, Norway; (M.J.); (J.H.A.)
| | - Johanna U. Ericson
- Research Group for Host Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, 9019 Tromsø, Norway; (E.M.); (V.K.D.)
- Correspondence: (E.J.); (J.U.E.)
| |
Collapse
|
17
|
Gurnani M, Chauhan A, Ranjan A, Tuli HS, Alkhanani MF, Haque S, Dhama K, Lal R, Jindal T. Filamentous Thermosensitive Mutant Z: An Appealing Target for Emerging Pathogens and a Trek on Its Natural Inhibitors. BIOLOGY 2022; 11:624. [PMID: 35625352 PMCID: PMC9138142 DOI: 10.3390/biology11050624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/18/2022] [Accepted: 04/01/2022] [Indexed: 12/14/2022]
Abstract
Antibiotic resistance is a major emerging issue in the health care sector, as highlighted by the WHO. Filamentous Thermosensitive mutant Z (Fts-Z) is gaining significant attention in the scientific community as a potential anti-bacterial target for fighting antibiotic resistance among several pathogenic bacteria. The Fts-Z plays a key role in bacterial cell division by allowing Z ring formation. Several in vitro and in silico experiments have demonstrated that inhibition of Fts-Z can lead to filamentous growth of the cells, and finally, cell death occurs. Many natural compounds that have successfully inhibited Fts-Z are also studied. This review article intended to highlight the structural-functional aspect of Fts-Z that leads to Z-ring formation and its contribution to the biochemistry and physiology of cells. The current trend of natural inhibitors of Fts-Z protein is also covered.
Collapse
Affiliation(s)
- Manisha Gurnani
- Amity Institute of Environmental Science, Amity University, Noida 201301, India;
| | - Abhishek Chauhan
- Amity Institute of Environmental Toxicology, Safety and Management, Amity University, Noida 201303, India;
| | - Anuj Ranjan
- Academy of Biology and Biotechnology, Southern Federal University, 344006 Rostov-on-Don, Russia
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Ambala 133207, India;
| | - Mustfa F. Alkhanani
- Emergency Service Department, College of Applied Sciences, AlMaarefa University, Riyadh 11597, Saudi Arabia;
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia;
- Faculty of Medicine, Görükle Campus, Bursa Uludağ University, Nilüfer, Bursa 16059, Turkey
| | - Kuldeep Dhama
- Division of Pathology, ICAR—Indian Veterinary Research Institute, Bareilly 243122, India;
| | - Rup Lal
- Department of Zoology, University of Delhi, Delhi 110021, India;
| | - Tanu Jindal
- Amity Institute of Environmental Toxicology, Safety and Management, Amity University, Noida 201303, India;
| |
Collapse
|
18
|
Gurnani M, Rath P, Chauhan A, Ranjan A, Ghosh A, Lal R, Mukerjee N, Aljarba NH, Alkahtani S, Rajput VD, Sushkova S, Prazdnova EV, Minkina T, Jindal T. Inhibition of Filamentous Thermosensitive Mutant-Z Protein in Bacillus subtilis by Cyanobacterial Bioactive Compounds. Molecules 2022; 27:1907. [PMID: 35335270 PMCID: PMC8948890 DOI: 10.3390/molecules27061907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 01/13/2023] Open
Abstract
Antibiotic resistance is one of the major growing concerns for public health. Conventional antibiotics act on a few predefined targets and, with time, several bacteria have developed resistance against a large number of antibiotics. The WHO has suggested that antibiotic resistance is at a crisis stage and identification of new antibiotics and targets could be the only approach to bridge the gap. Filamentous Temperature Sensitive-Mutant Z (Fts-Z) is one of the promising and less explored antibiotic targets. It is a highly conserved protein and plays a key role in bacterial cell division by introducing a cytokinetic Z-ring formation. In the present article, the potential of over 165 cyanobacterial compounds with reported antibiotic activity against the catalytic core domain in the Fts-Z protein of the Bacillus subtilis was studied. The identified cyanobacterial compounds were screened using the GLIDE module of Maestro v-2019-2 followed by 100-ns molecular dynamics (MD) simulation. Ranking of the potential compound was performed using dock score and MMGBSA based free energy. The study reported that the docking score of aphanorphine (-6.010 Kcalmol-1) and alpha-dimorphecolic acid (ADMA) (-6.574 Kcalmol-1) showed significant role with respect to the reported potential inhibitor PC190723 (-4.135 Kcalmol-1). A 100 ns MD simulation infers that Fts-Z ADMA complex has a stable conformation throughout the progress of the simulation. Both the compounds, i.e., ADMA and Aphanorphine, were further considered for In-vitro validation by performing anti-bacterial studies against B. subtilis by agar well diffusion method. The results obtained through In-vitro studies confirm that ADMA, a small molecule of cyanobacterial origin, is a potential compound with an antibacterial activity that may act by inhibiting the novel target Fts-Z and could be a great drug candidate for antibiotic development.
Collapse
Affiliation(s)
- Manisha Gurnani
- Amity Institute of Environmental Sciences, Amity University, Noida 201301, India; (M.G.); (P.R.)
| | - Prangya Rath
- Amity Institute of Environmental Sciences, Amity University, Noida 201301, India; (M.G.); (P.R.)
| | - Abhishek Chauhan
- Amity Institute of Environment Toxicology and Safety Management, Amity University, Noida 201303, India;
| | - Anuj Ranjan
- Academy of Biology and Biotechnology, Southern Federal University, 344006 Rostov-on-Don, Russia; (V.D.R.); (S.S.); (E.V.P.); (T.M.)
| | - Arabinda Ghosh
- Department of Botany, Microbiology Division, Guwahati University, Guwahati 781014, India;
| | - Rup Lal
- Department of Zoology, University of Delhi, New Delhi 110007, India;
| | - Nobendu Mukerjee
- Department of Microbiology, Ramakrishna Mission Vivekananda Centenary College, Kolkata 700118, India;
- Department of Health Sciences, Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia
| | - Nada H. Aljarba
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
| | - Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Vishnu D. Rajput
- Academy of Biology and Biotechnology, Southern Federal University, 344006 Rostov-on-Don, Russia; (V.D.R.); (S.S.); (E.V.P.); (T.M.)
| | - Svetlana Sushkova
- Academy of Biology and Biotechnology, Southern Federal University, 344006 Rostov-on-Don, Russia; (V.D.R.); (S.S.); (E.V.P.); (T.M.)
| | - Evgenya V. Prazdnova
- Academy of Biology and Biotechnology, Southern Federal University, 344006 Rostov-on-Don, Russia; (V.D.R.); (S.S.); (E.V.P.); (T.M.)
| | - Tatiana Minkina
- Academy of Biology and Biotechnology, Southern Federal University, 344006 Rostov-on-Don, Russia; (V.D.R.); (S.S.); (E.V.P.); (T.M.)
| | - Tanu Jindal
- Amity Institute of Environment Toxicology and Safety Management, Amity University, Noida 201303, India;
| |
Collapse
|
19
|
Dat TD, Viet ND, Thanh VH, Linh NTT, Ngan NTK, Nam HM, Phong MT, Hieu NH. Optimization of Triterpenoid Extraction from
Ganoderma lucidum
by Ethanol‐Modified Supercritical Carbon Dioxide andthe Biological Properties of the Extract. ChemistrySelect 2022. [DOI: 10.1002/slct.202103444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Tran Do Dat
- VNU-HCMC, Key Laboratory of Chemical Engineering and Petroleum Processing (Key CEPP Lab) Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, District 10 Ho Chi Minh City Vietnam
- Faculty of Chemical Engineering Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, Ward 14, District 10, Ho Chi Minh City Vietnam
- Vietnam National University Ho Chi Minh City (VNU-HCM), Linh Trung Ward, Thu Duc City Ho Chi Minh City Vietnam
| | - Nguyen Duc Viet
- VNU-HCMC, Key Laboratory of Chemical Engineering and Petroleum Processing (Key CEPP Lab) Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, District 10 Ho Chi Minh City Vietnam
- Faculty of Chemical Engineering Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, Ward 14, District 10, Ho Chi Minh City Vietnam
- Vietnam National University Ho Chi Minh City (VNU-HCM), Linh Trung Ward, Thu Duc City Ho Chi Minh City Vietnam
| | - Vuong Hoai Thanh
- VNU-HCMC, Key Laboratory of Chemical Engineering and Petroleum Processing (Key CEPP Lab) Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, District 10 Ho Chi Minh City Vietnam
- Faculty of Chemical Engineering Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, Ward 14, District 10, Ho Chi Minh City Vietnam
- Vietnam National University Ho Chi Minh City (VNU-HCM), Linh Trung Ward, Thu Duc City Ho Chi Minh City Vietnam
| | - Ngo Thi Thuy Linh
- VNU-HCMC, Key Laboratory of Chemical Engineering and Petroleum Processing (Key CEPP Lab) Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, District 10 Ho Chi Minh City Vietnam
- Faculty of Chemical Engineering Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, Ward 14, District 10, Ho Chi Minh City Vietnam
- Vietnam National University Ho Chi Minh City (VNU-HCM), Linh Trung Ward, Thu Duc City Ho Chi Minh City Vietnam
| | - Nguyen Thi Kim Ngan
- VNU-HCMC, Key Laboratory of Chemical Engineering and Petroleum Processing (Key CEPP Lab) Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, District 10 Ho Chi Minh City Vietnam
- Faculty of Chemical Engineering Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, Ward 14, District 10, Ho Chi Minh City Vietnam
- Vietnam National University Ho Chi Minh City (VNU-HCM), Linh Trung Ward, Thu Duc City Ho Chi Minh City Vietnam
| | - Hoang Minh Nam
- VNU-HCMC, Key Laboratory of Chemical Engineering and Petroleum Processing (Key CEPP Lab) Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, District 10 Ho Chi Minh City Vietnam
- Faculty of Chemical Engineering Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, Ward 14, District 10, Ho Chi Minh City Vietnam
- Vietnam National University Ho Chi Minh City (VNU-HCM), Linh Trung Ward, Thu Duc City Ho Chi Minh City Vietnam
| | - Mai Thanh Phong
- VNU-HCMC, Key Laboratory of Chemical Engineering and Petroleum Processing (Key CEPP Lab) Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, District 10 Ho Chi Minh City Vietnam
- Faculty of Chemical Engineering Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, Ward 14, District 10, Ho Chi Minh City Vietnam
- Vietnam National University Ho Chi Minh City (VNU-HCM), Linh Trung Ward, Thu Duc City Ho Chi Minh City Vietnam
| | - Nguyen Huu Hieu
- VNU-HCMC, Key Laboratory of Chemical Engineering and Petroleum Processing (Key CEPP Lab) Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, District 10 Ho Chi Minh City Vietnam
- Faculty of Chemical Engineering Ho Chi Minh City University of Technology (HCMUT) 268 Ly Thuong Kiet Street, Ward 14, District 10, Ho Chi Minh City Vietnam
- Vietnam National University Ho Chi Minh City (VNU-HCM), Linh Trung Ward, Thu Duc City Ho Chi Minh City Vietnam
| |
Collapse
|
20
|
Du RL, Sun N, Fung YH, Zheng YY, Chen YW, Chan PH, Wong WL, Wong KY. Discovery of FtsZ inhibitors by virtual screening as antibacterial agents and study of the inhibition mechanism. RSC Med Chem 2022; 13:79-89. [PMID: 35224498 PMCID: PMC8792978 DOI: 10.1039/d1md00249j] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/07/2021] [Indexed: 09/29/2023] Open
Abstract
Inhibition of bacterial cell division is a novel mechanistic action in the development of new antimicrobial agents. The FtsZ protein is an important antimicrobial drug target because of its essential role in bacterial cell division. In the present study, potential inhibitors of FtsZ were identified by virtual screening followed by in vivo and in vitro bioassays. One of the candidates, Dacomitinib (S2727), shows for the first time its potent inhibitory activity against the MRSA strains. The binding mode of Dacomitinib in FtsZ was analyzed by docking, and Asp199 and Thr265 are thought to be essential residues involved in the interactions.
Collapse
Affiliation(s)
- Ruo-Lan Du
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| | - Ning Sun
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| | - Yik-Hong Fung
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| | - Yuan-Yuan Zheng
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| | - Yu-Wei Chen
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| | - Pak-Ho Chan
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| | - Kwok-Yin Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| |
Collapse
|
21
|
Rodrigo S, García-Latorre C, Santamaria O. Metabolites Produced by Fungi against Fungal Phytopathogens: Review, Implementation and Perspectives. PLANTS (BASEL, SWITZERLAND) 2021; 11:81. [PMID: 35009084 PMCID: PMC8747711 DOI: 10.3390/plants11010081] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 01/06/2023]
Abstract
Many fungi, especially endophytes, have been found to produce multiple benefits in their plant hosts, with many of these benefits associated with the protection of plants against fungal diseases. This fact could be used in the development of new bio-products that could gradually reduce the need for chemical fungicides, which have been associated with multiple health and environmental problems. However, the utilization of the living organism may present several issues, such as an inconsistency in the results obtained and more complicated management and application, as fungal species are highly influenced by environmental conditions, the type of relationship with the plant host and interaction with other microorganisms. These issues could be addressed by using the bioactive compounds produced by the fungus, in cases where they were responsible for positive effects, instead of the living organism. Multiple bioactive compounds produced by fungal species, especially endophytes, with antifungal properties have been previously reported in the literature. However, despite the large amount of these metabolites and their potential, extensive in-field application on a large scale has not yet been implemented. In the present review, the main aspects explaining this limited implementation are analyzed, and the present and future perspectives for its development are discussed.
Collapse
Affiliation(s)
- Sara Rodrigo
- Department of Agronomy and Forest Environment Engineering, University of Extremadura, Avda, Adolfo Suárez s/n, 06007 Badajoz, Spain; (S.R.); (C.G.-L.)
| | - Carlos García-Latorre
- Department of Agronomy and Forest Environment Engineering, University of Extremadura, Avda, Adolfo Suárez s/n, 06007 Badajoz, Spain; (S.R.); (C.G.-L.)
| | - Oscar Santamaria
- Department of Construction and Agronomy, University of Salamanca, Avda, Cardenal Cisneros 34, 49029 Zamora, Spain
| |
Collapse
|
22
|
Privalsky TM, Soohoo AM, Wang J, Walsh CT, Wright GD, Gordon EM, Gray NS, Khosla C. Prospects for Antibacterial Discovery and Development. J Am Chem Soc 2021; 143:21127-21142. [PMID: 34860516 PMCID: PMC8855840 DOI: 10.1021/jacs.1c10200] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The rising prevalence of multidrug-resistant bacteria is an urgent health crisis that can only be countered through renewed investment in the discovery and development of antibiotics. There is no panacea for the antibacterial resistance crisis; instead, a multifaceted approach is called for. In this Perspective we make the case that, in the face of evolving clinical needs and enabling technologies, numerous validated antibacterial targets and associated lead molecules deserve a second look. At the same time, many worthy targets lack good leads despite harboring druggable active sites. Creative and inspired techniques buoy discovery efforts; while soil screening efforts frequently lead to antibiotic rediscovery, researchers have found success searching for new antibiotic leads by studying underexplored ecological niches or by leveraging the abundance of available data from genome mining efforts. The judicious use of "polypharmacology" (i.e., the ability of a drug to alter the activities of multiple targets) can also provide new opportunities, as can the continued search for inhibitors of resistance enzymes with the capacity to breathe new life into old antibiotics. We conclude by highlighting available pharmacoeconomic models for antibacterial discovery and development while making the case for new ones.
Collapse
Affiliation(s)
- Thomas M. Privalsky
- Department of Chemistry, Stanford University, Stanford, CA 94305, United States
| | - Alexander M. Soohoo
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, United States
| | - Jinhua Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 United States
| | - Christopher T. Walsh
- Department of Chemistry, Stanford University, Stanford, CA 94305, United States
- Stanford ChEM-H, Stanford University, Stanford, CA 94305, United States
| | - Gerard D. Wright
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Eric M. Gordon
- Stanford ChEM-H, Stanford University, Stanford, CA 94305, United States
- Department of Medicine, Stanford University, Stanford, CA 94305, United States
| | - Nathanael S. Gray
- Stanford ChEM-H, Stanford University, Stanford, CA 94305, United States
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, United States
| | - Chaitan Khosla
- Department of Chemistry, Stanford University, Stanford, CA 94305, United States
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, United States
- Stanford ChEM-H, Stanford University, Stanford, CA 94305, United States
| |
Collapse
|
23
|
Graham CLB, Newman H, Gillett FN, Smart K, Briggs N, Banzhaf M, Roper DI. A Dynamic Network of Proteins Facilitate Cell Envelope Biogenesis in Gram-Negative Bacteria. Int J Mol Sci 2021; 22:12831. [PMID: 34884635 PMCID: PMC8657477 DOI: 10.3390/ijms222312831] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 01/01/2023] Open
Abstract
Bacteria must maintain the ability to modify and repair the peptidoglycan layer without jeopardising its essential functions in cell shape, cellular integrity and intermolecular interactions. A range of new experimental techniques is bringing an advanced understanding of how bacteria regulate and achieve peptidoglycan synthesis, particularly in respect of the central role played by complexes of Sporulation, Elongation or Division (SEDs) and class B penicillin-binding proteins required for cell division, growth and shape. In this review we highlight relationships implicated by a bioinformatic approach between the outer membrane, cytoskeletal components, periplasmic control proteins, and cell elongation/division proteins to provide further perspective on the interactions of these cell division, growth and shape complexes. We detail the network of protein interactions that assist in the formation of peptidoglycan and highlight the increasingly dynamic and connected set of protein machinery and macrostructures that assist in creating the cell envelope layers in Gram-negative bacteria.
Collapse
Affiliation(s)
- Chris L. B. Graham
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK; (C.L.B.G.); (H.N.); (F.N.G.); (K.S.); (N.B.)
| | - Hector Newman
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK; (C.L.B.G.); (H.N.); (F.N.G.); (K.S.); (N.B.)
| | - Francesca N. Gillett
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK; (C.L.B.G.); (H.N.); (F.N.G.); (K.S.); (N.B.)
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Katie Smart
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK; (C.L.B.G.); (H.N.); (F.N.G.); (K.S.); (N.B.)
| | - Nicholas Briggs
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK; (C.L.B.G.); (H.N.); (F.N.G.); (K.S.); (N.B.)
| | - Manuel Banzhaf
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - David I. Roper
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK; (C.L.B.G.); (H.N.); (F.N.G.); (K.S.); (N.B.)
| |
Collapse
|
24
|
Li H, Shu S, Kalaitzis JA, Shang Z, Vuong D, Crombie A, Lacey E, Piggott AM, Chooi YH. Genome Mining of Aspergillus hancockii Unearths Cryptic Polyketide Hancockinone A Featuring a Prenylated 6/6/6/5 Carbocyclic Skeleton. Org Lett 2021; 23:8789-8793. [PMID: 34747627 DOI: 10.1021/acs.orglett.1c03283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Activation of a cryptic polyketide synthase gene cluster hkn from Aspergillus hancockii via overexpression of the gene-cluster-specific transcription factor HknR led to the discovery of a novel polycyclic metabolite, which we named hancockinone A. The compound features an unprecedented prenylated 6/6/6/5 tetracarbocyclic skeleton and shows moderate antibacterial activity. Heterologous expression, substrate feeding, and in vitro assays confirmed the role of cytochrome P450 HknE in constructing the five-membered ring in hancockinone A from the precursor neosartoricin B.
Collapse
Affiliation(s)
- Hang Li
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Si Shu
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - John A Kalaitzis
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
- Microbial Screening Technologies Pty. Ltd., Smithfield, NSW 2164, Australia
| | - Zhuo Shang
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Daniel Vuong
- Microbial Screening Technologies Pty. Ltd., Smithfield, NSW 2164, Australia
| | - Andrew Crombie
- Microbial Screening Technologies Pty. Ltd., Smithfield, NSW 2164, Australia
| | - Ernest Lacey
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
- Microbial Screening Technologies Pty. Ltd., Smithfield, NSW 2164, Australia
| | - Andrew M Piggott
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Yit-Heng Chooi
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
25
|
Jenssen M, Rainsford P, Juskewitz E, Andersen JH, Hansen EH, Isaksson J, Rämä T, Hansen KØ. Lulworthinone, a New Dimeric Naphthopyrone From a Marine Fungus in the Family Lulworthiaceae With Antibacterial Activity Against Clinical Methicillin-Resistant Staphylococcus aureus Isolates. Front Microbiol 2021; 12:730740. [PMID: 34659158 PMCID: PMC8517231 DOI: 10.3389/fmicb.2021.730740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/06/2021] [Indexed: 11/13/2022] Open
Abstract
The emergence of drug-resistant bacteria is increasing rapidly in all parts of the world, and the need for new antibiotics is urgent. In our continuous search for new antimicrobial molecules from under-investigated Arctic marine microorganisms, a marine fungus belonging to the family Lulworthiaceae (Lulworthiales, Sordariomycetes, and Ascomycota) was studied. The fungus was isolated from driftwood, cultivated in liquid medium, and studied for its potential for producing antibacterial compounds. Through bioactivity-guided isolation, a novel sulfated biarylic naphtho-α-pyrone dimer was isolated, and its structure was elucidated by spectroscopic methods, including 1D and 2D NMR and HRMS. The compound, named lulworthinone (1), showed antibacterial activity against reference strains of Staphylococcus aureus and Streptococcus agalactiae, as well as several clinical MRSA isolates with MICs in the 1.56-6.25 μg/ml range. The compound also had antiproliferative activity against human melanoma, hepatocellular carcinoma, and non-malignant lung fibroblast cell lines, with IC50 values of 15.5, 27, and 32 μg/ml, respectively. Inhibition of bacterial biofilm formation was observed, but no eradication of established biofilm could be detected. No antifungal activity was observed against Candida albicans. During the isolation of 1, the compound was observed to convert into a structural isomer, 2, under acidic conditions. As 1 and 2 have high structural similarity, NMR data acquired for 2 were used to aid in the structure elucidation of 1. To the best of our knowledge, lulworthinone (1) represents the first new bioactive secondary metabolite isolated from the marine fungal order Lulworthiales.
Collapse
Affiliation(s)
- Marte Jenssen
- Marbio, The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT the Arctic University of Norway, Tromsø, Norway
| | - Philip Rainsford
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, Tromsø, Norway
| | - Eric Juskewitz
- Research Group for Host Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, Tromsø, Norway
| | - Jeanette H. Andersen
- Marbio, The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT the Arctic University of Norway, Tromsø, Norway
| | - Espen H. Hansen
- Marbio, The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT the Arctic University of Norway, Tromsø, Norway
| | - Johan Isaksson
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, Tromsø, Norway
| | - Teppo Rämä
- Marbio, The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT the Arctic University of Norway, Tromsø, Norway
| | - Kine Ø. Hansen
- Marbio, The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT the Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
26
|
Kumar M, Mathur T, Barman TK, Chaira T, Kumar R, Joshi V, Pandya M, Sharma L, Fujii K, Bandgar M, Jadhav B, Bambal R, Upadhyay D, Masuda N, Verma AK, Bhatnagar PK. Novel FtsZ inhibitor with potent activity against Staphylococcus aureus. J Antimicrob Chemother 2021; 76:2867-2874. [PMID: 34383913 DOI: 10.1093/jac/dkab270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 06/29/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES FtsZ is an essential bacterial protein and an unexplored target for the development of antibacterial drugs. The development of a novel inhibitor targeting FtsZ offers a potential opportunity to combat drug resistance. DS01750413, a new derivative of PC190723, is a novel FtsZ inhibitor with improved in vitro and in vivo activity. The objective of this study was to investigate the efficacy of DS01750413 against Staphylococcus spp., including MRSA, in in vitro and in vivo models. METHODS In vitro activities of DS01750413 and standard-of-care antibiotics were evaluated against clinical isolates of Gram-positive pathogens. The in vivo efficacy was evaluated in a murine systemic infection model caused by MRSA. RESULTS DS01750413 showed potent in vitro activity against MRSA clinical isolates with MIC ranges of 0.5-1 mg/L and also demonstrated concentration-dependent bactericidal killing. In the murine bacteraemia infection model of MRSA, treatment with DS01750413 resulted in prolonged survival of animals compared with placebo-treated animals and exhibited a significant reduction in the bacterial load in liver, spleen, lungs and kidneys. CONCLUSIONS DS01750413 showed encouraging in vitro and in vivo activity against MRSA. As a novel chemical class, DS01750413 has the potential to become clinically viable antibiotics to address the drug resistance problem by its unique novel targeting mechanism of action.
Collapse
Affiliation(s)
- Manoj Kumar
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| | - Tarun Mathur
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| | - Tarani Kanta Barman
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| | - Tridib Chaira
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| | - Ram Kumar
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| | - Vattan Joshi
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| | - Manisha Pandya
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| | - Lalima Sharma
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| | - Kunihiko Fujii
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| | - Mahadev Bandgar
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| | - Balasaheb Jadhav
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| | - Ramesh Bambal
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| | - Dilip Upadhyay
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| | - Nobuhisa Masuda
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| | - Ashwani Kumar Verma
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| | - Pradip Kumar Bhatnagar
- Daiichi Sankyo India Pharma Private Limited, Village Sarhaul, Sector-18, Udyog Vihar Industrial Area, Gurgaon 122 015, Haryana, India
| |
Collapse
|
27
|
Milani G, Cavalluzzi MM, Solidoro R, Salvagno L, Quintieri L, Di Somma A, Rosato A, Corbo F, Franchini C, Duilio A, Caputo L, Habtemariam S, Lentini G. Molecular Simplification of Natural Products: Synthesis, Antibacterial Activity, and Molecular Docking Studies of Berberine Open Models. Biomedicines 2021; 9:452. [PMID: 33922200 PMCID: PMC8146520 DOI: 10.3390/biomedicines9050452] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 02/08/2023] Open
Abstract
Berberine, the main bioactive component of many medicinal plants belonging to various genera such as Berberis, Coptis, and Hydrastis is a multifunctional compound. Among the numerous interesting biological properties of berberine is broad antimicrobial activity including a range of Gram-positive and Gram-negative bacteria. With the aim of identifying berberine analogues possibly endowed with higher lead-likeness and easier synthetic access, the molecular simplification approach was applied to the secondary metabolite and a series of analogues were prepared and screened for their antimicrobial activity against Gram-positive and Gram-negative bacterial test species. Rewardingly, the berberine simplified analogues displayed 2-20-fold higher potency with respect to berberine. Since our berberine simplified analogues may be easily synthesized and are characterized by lower molecular weight than the parent compound, they are further functionalizable and should be more suitable for oral administration. Molecular docking simulations suggested FtsZ, a well-known protein involved in bacterial cell division, as a possible target.
Collapse
Affiliation(s)
- Gualtiero Milani
- Department of Pharmacy–Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona n. 4, 70126 Bari, Italy; (G.M.); (R.S.); (L.S.); (A.R.); (F.C.); (C.F.); (G.L.)
| | - Maria Maddalena Cavalluzzi
- Department of Pharmacy–Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona n. 4, 70126 Bari, Italy; (G.M.); (R.S.); (L.S.); (A.R.); (F.C.); (C.F.); (G.L.)
| | - Roberta Solidoro
- Department of Pharmacy–Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona n. 4, 70126 Bari, Italy; (G.M.); (R.S.); (L.S.); (A.R.); (F.C.); (C.F.); (G.L.)
| | - Lara Salvagno
- Department of Pharmacy–Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona n. 4, 70126 Bari, Italy; (G.M.); (R.S.); (L.S.); (A.R.); (F.C.); (C.F.); (G.L.)
| | - Laura Quintieri
- Institute of Sciences of Food Production (CNR-ISPA) National Council of Research, Via G. Amendola, 122/O, 70126 Bari, Italy; (L.Q.); (L.C.)
| | - Angela Di Somma
- Department of Chemical Sciences, University of Naples “Federico II” Via Cinthia 4, 80126 Napoli, Italy; (A.D.S.); (A.D.)
| | - Antonio Rosato
- Department of Pharmacy–Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona n. 4, 70126 Bari, Italy; (G.M.); (R.S.); (L.S.); (A.R.); (F.C.); (C.F.); (G.L.)
| | - Filomena Corbo
- Department of Pharmacy–Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona n. 4, 70126 Bari, Italy; (G.M.); (R.S.); (L.S.); (A.R.); (F.C.); (C.F.); (G.L.)
| | - Carlo Franchini
- Department of Pharmacy–Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona n. 4, 70126 Bari, Italy; (G.M.); (R.S.); (L.S.); (A.R.); (F.C.); (C.F.); (G.L.)
| | - Angela Duilio
- Department of Chemical Sciences, University of Naples “Federico II” Via Cinthia 4, 80126 Napoli, Italy; (A.D.S.); (A.D.)
| | - Leonardo Caputo
- Institute of Sciences of Food Production (CNR-ISPA) National Council of Research, Via G. Amendola, 122/O, 70126 Bari, Italy; (L.Q.); (L.C.)
| | - Solomon Habtemariam
- Pharmacognosy Research Laboratories & Herbal Analysis Services, University of Greenwich, Chatham-Maritime, Kent ME4 4TB, UK;
| | - Giovanni Lentini
- Department of Pharmacy–Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona n. 4, 70126 Bari, Italy; (G.M.); (R.S.); (L.S.); (A.R.); (F.C.); (C.F.); (G.L.)
| |
Collapse
|
28
|
Takahashi H, Umemura M, Ninomiya A, Kusuya Y, Shimizu M, Urayama SI, Watanabe A, Kamei K, Yaguchi T, Hagiwara D. Interspecies Genomic Variation and Transcriptional Activeness of Secondary Metabolism-Related Genes in Aspergillus Section Fumigati. FRONTIERS IN FUNGAL BIOLOGY 2021; 2:656751. [PMID: 37744138 PMCID: PMC10512231 DOI: 10.3389/ffunb.2021.656751] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/23/2021] [Indexed: 09/26/2023]
Abstract
Filamentous fungi produce various bioactive compounds that are biosynthesized by sets of proteins encoded in biosynthesis gene clusters (BGCs). For an unknown reason, many BGCs are transcriptionally silent in laboratory conditions, which has hampered the discovery of novel fungal compounds. The transcriptional reactiveness of fungal secondary metabolism is not fully understood. To gain the comprehensive view, we conducted comparative genomic and transcriptomic analyses of nine closely-related species of Aspergillus section Fumigati (A. fumigatus, A. fumigatiaffinis, A. novofumigatus, A. thermomutatus, A. viridinutans, A. pseudoviridinutans, A. lentulus, A. udagawae, and Neosartorya fischeri). For expanding our knowledge, we newly sequenced genomes of A. viridinutans and A. pseudoviridinutans, and reassembled and reannotated the previously released genomes of A. lentulus and A. udagawae. Between 34 and 84 secondary metabolite (SM) backbone genes were identified in the genomes of these nine respective species, with 8.7-51.2% being unique to the species. A total of 247 SM backbone gene types were identified in the nine fungi. Ten BGCs are shared by all nine species. Transcriptomic analysis using A. fumigatus, A. lentulus, A. udagawae, A. viridinutans, and N. fischeri was conducted to compare expression levels of all SM backbone genes in four different culture conditions; 32-83% of SM backbone genes in these species were not expressed in the tested conditions, which reconfirmed that large part of fungal SM genes are hard to be expressed. The species-unique SM genes of the five species were expressed with lower frequency (18.8% in total) than the SM genes that are conserved in all five species (56%). These results suggest that the expression tendency of BGCs is correlated with their interspecies distribution pattern. Our findings increase understanding of the evolutionary processes associated with the regulation of fungal secondary metabolism.
Collapse
Affiliation(s)
- Hiroki Takahashi
- Medical Mycology Research Center, Chiba University, Chiba, Japan
- Molecular Chirality Research Center, Chiba University, Chiba, Japan
- Plant Molecular Science Center, Chiba University, Chiba, Japan
| | - Maiko Umemura
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Akihiro Ninomiya
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Yoko Kusuya
- Medical Mycology Research Center, Chiba University, Chiba, Japan
| | - Masaaki Shimizu
- Department of Biology, Faculty of Science, Chiba University, Chiba, Japan
| | - Syun-ichi Urayama
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
- Microbiology Research Center for Sustainability, University of Tsukuba, Tsukuba, Japan
| | - Akira Watanabe
- Medical Mycology Research Center, Chiba University, Chiba, Japan
| | - Katsuhiko Kamei
- Medical Mycology Research Center, Chiba University, Chiba, Japan
| | - Takashi Yaguchi
- Medical Mycology Research Center, Chiba University, Chiba, Japan
| | - Daisuke Hagiwara
- Medical Mycology Research Center, Chiba University, Chiba, Japan
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
- Microbiology Research Center for Sustainability, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
29
|
Hagee D, Abu Hardan A, Botero J, Arnone JT. Genomic clustering within functionally related gene families in Ascomycota fungi. Comput Struct Biotechnol J 2020; 18:3267-3277. [PMID: 33209211 PMCID: PMC7653285 DOI: 10.1016/j.csbj.2020.10.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/15/2020] [Accepted: 10/17/2020] [Indexed: 12/17/2022] Open
Abstract
Multiple mechanisms collaborate for proper regulation of gene expression. One layer of this regulation is through the clustering of functionally related genes at discrete loci throughout the genome. This phenomenon occurs extensively throughout Ascomycota fungi and is an organizing principle for many gene families whose proteins participate in diverse molecular functions throughout the cell. Members of this phylum include organisms that serve as model systems and those of interest medically, pharmaceutically, and for industrial and biotechnological applications. In this review, we discuss the prevalence of functional clustering through a broad range of organisms within the phylum. Position effects on transcription, genomic locations of clusters, transcriptional regulation of clusters, and selective pressures contributing to the formation and maintenance of clusters are addressed, as are common methods to identify and characterize clusters.
Collapse
Affiliation(s)
- Danielle Hagee
- Department of Biology, William Paterson University, Wayne, NJ 07470, USA
| | - Ahmad Abu Hardan
- Department of Biology, William Paterson University, Wayne, NJ 07470, USA
| | - Juan Botero
- Department of Biology, William Paterson University, Wayne, NJ 07470, USA
| | - James T. Arnone
- Department of Biology, William Paterson University, Wayne, NJ 07470, USA
| |
Collapse
|
30
|
Su M, Zhao C, Li D, Cao J, Ju Z, Kim EL, Jung YS, Jung JH. Viriditoxin Stabilizes Microtubule Polymers in SK-OV-3 Cells and Exhibits Antimitotic and Antimetastatic Potential. Mar Drugs 2020; 18:md18090445. [PMID: 32867174 PMCID: PMC7551567 DOI: 10.3390/md18090445] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 01/08/2023] Open
Abstract
Microtubules play a crucial role in mitosis and are attractive targets for cancer therapy. Recently, we isolated viriditoxin, a cytotoxic and antibacterial compound, from a marine fungus Paecilomyces variotii. Viriditoxin has been reported to inhibit the polymerization of bacterial FtsZ, a tubulin-like GTPase that plays an essential role in bacterial cell division. Given the close structural homology between FtsZ and tubulin, we investigated the potential antimitotic effects of viriditoxin on human cancer cells. Viriditoxin, like paclitaxel, enhanced tubulin polymerization and stabilized microtubule polymers, thereby perturbing mitosis in the SK-OV-3 cell line. However, the morphology of the stabilized microtubules was different from that induced by paclitaxel, indicating subtle differences in the mode of action of these compounds. Microtubule dynamics are also essential in cell movement, and viriditoxin repressed migration and colony formation ability of SK-OV-3 cells. Based on these results, we propose that viriditoxin interrupts microtubule dynamics, thus leading to antimitotic and antimetastatic activities.
Collapse
Affiliation(s)
- Mingzhi Su
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
| | - Changhao Zhao
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
| | - Dandan Li
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
| | - Jiafu Cao
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
| | - Zhiran Ju
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
| | - Eun La Kim
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
| | - Young-Suk Jung
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
| | - Jee H. Jung
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (M.S.); (C.Z.); (D.L.); (J.C.); (Z.J.); (E.L.K.); (Y.-S.J.)
- Correspondence:
| |
Collapse
|
31
|
Ur Rahman M, Wang P, Wang N, Chen Y. A key bacterial cytoskeletal cell division protein FtsZ as a novel therapeutic antibacterial drug target. Bosn J Basic Med Sci 2020; 20:310-318. [PMID: 32020845 PMCID: PMC7416170 DOI: 10.17305/bjbms.2020.4597] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 01/25/2020] [Indexed: 12/18/2022] Open
Abstract
Nowadays, the emergence of multidrug-resistant bacterial strains initiates the urgent need for the elucidation of the new drug targets for the discovery of antimicrobial drugs. Filamenting temperature-sensitive mutant Z (FtsZ), a eukaryotic tubulin homolog, is a GTP-dependent prokaryotic cytoskeletal protein and is conserved among most bacterial strains. In vitro studies revealed that FtsZ self-assembles into dynamic protofilaments or bundles and forms a dynamic Z-ring at the center of the cell in vivo, leading to septation and consequent cell division. Speculations on the ability of FtsZ in the blockage of cell division make FtsZ a highly attractive target for developing novel antibiotics. Researchers have been working on synthetic molecules and natural products as inhibitors of FtsZ. Accumulating data suggest that FtsZ may provide the platform for the development of novel antibiotics. In this review, we summarize recent advances in the properties of FtsZ protein and bacterial cell division, as well as in the development of FtsZ inhibitors.
Collapse
Affiliation(s)
- Mujeeb Ur Rahman
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| | - Ping Wang
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Na Wang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| | - Yaodong Chen
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| |
Collapse
|
32
|
Chung PY. Novel targets of pentacyclic triterpenoids in Staphylococcus aureus: A systematic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 73:152933. [PMID: 31103429 DOI: 10.1016/j.phymed.2019.152933] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 04/15/2019] [Accepted: 04/16/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Staphylococcus aureus is an important pathogen both in community-acquired and healthcare-associated infections, and has successfully evolved numerous strategies for resisting the action to practically all antibiotics. Resistance to methicillin is now widely described in the community setting (CMRSA), thus the development of new drugs or alternative therapies is urgently necessary. Plants and their secondary metabolites have been a major alternative source in providing structurally diverse bioactive compounds as potential therapeutic agents for the treatment of bacterial infections. One of the classes of natural secondary metabolites from plants with the most bioactive compounds are the triterpenoids, which comprises structurally diverse organic compounds. In nature, triterpenoids are often found as tetra- or penta-cyclic structures. AIM This review highlights the anti-staphylococcal activities of pentacyclic triterpenoids, particularly α-amyrin (AM), betulinic acid (BA) and betulinaldehyde (BE). These compounds are based on a 30-carbon skeleton comprising five six-membered rings (ursanes and lanostanes) or four six-membered rings and one five-membered ring (lupanes and hopanes). METHODS Electronic databases such as ScienceDirect, PubMed and Scopus were used to search scientific contributions until March 2018, using relevant keywords. Literature focusing on the antimicrobial and antibiofilms of effects of pentacyclic triterpenoids on S. aureus were identified and summarized. RESULTS Pentacyclic triterpenoids can be divided into three representative classes, namely ursane, lupane and oleananes. This class of compounds have been shown to exhibit analgesic, immunomodulatory, anti-inflammatory, anticancer, antioxidant, antifungal and antibacterial activities. In studies of the antimicrobial activities and targets of AM, BA and BE in sensitive and multidrug-resistant S. aureus, these compounds acted synergistically and have different targets from the conventional antibiotics. CONCLUSION The inhibitory mechanisms of S. aureus in novel targets and pathways should stimulate further researches to develop AM, BA and BE as therapeutic agents for infections caused by S. aureus. Continued efforts to identify and exploit synergistic combinations by the three compounds and peptidoglycan inhibitors, are also necessary as alternative treatment options for S. aureus infections.
Collapse
Affiliation(s)
- Pooi Yin Chung
- Department of Pathology, School of Medicine, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia.
| |
Collapse
|
33
|
Silber N, Matos de Opitz CL, Mayer C, Sass P. Cell division protein FtsZ: from structure and mechanism to antibiotic target. Future Microbiol 2020; 15:801-831. [DOI: 10.2217/fmb-2019-0348] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Antimicrobial resistance to virtually all clinically applied antibiotic classes severely limits the available options to treat bacterial infections. Hence, there is an urgent need to develop and evaluate new antibiotics and targets with resistance-breaking properties. Bacterial cell division has emerged as a new antibiotic target pathway to counteract multidrug-resistant pathogens. New approaches in antibiotic discovery and bacterial cell biology helped to identify compounds that either directly interact with the major cell division protein FtsZ, thereby perturbing the function and dynamics of the cell division machinery, or affect the structural integrity of FtsZ by inducing its degradation. The impressive antimicrobial activities and resistance-breaking properties of certain compounds validate the inhibition of bacterial cell division as a promising strategy for antibiotic intervention.
Collapse
Affiliation(s)
- Nadine Silber
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology & Infection Medicine, University of Tübingen, Auf der Morgenstelle 28, Tübingen 72076, Germany
| | - Cruz L Matos de Opitz
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology & Infection Medicine, University of Tübingen, Auf der Morgenstelle 28, Tübingen 72076, Germany
| | - Christian Mayer
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology & Infection Medicine, University of Tübingen, Auf der Morgenstelle 28, Tübingen 72076, Germany
| | - Peter Sass
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology & Infection Medicine, University of Tübingen, Auf der Morgenstelle 28, Tübingen 72076, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, Tübingen 72076, Germany
| |
Collapse
|
34
|
Chegeni TN, Fakhar M. Promising Role of Wolbachia as Anti-parasitic Drug Target and Eco-Friendly Biocontrol Agent. ACTA ACUST UNITED AC 2020; 14:69-79. [PMID: 30747079 DOI: 10.2174/1574891x14666190211162403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/08/2019] [Accepted: 01/12/2019] [Indexed: 01/13/2023]
Abstract
BACKGROUND Wolbachia is the most common endosymbiotic bacteria in insectborne parasites and it is the most common reproductive parasite in the world. Wolbachia has been found worldwide in numerous arthropod and parasite species, including insects, terrestrial isopods, spiders, mites and filarial nematodes. There is a complicated relationship between Wolbachia and its hosts and in some cases, they create a mutual relationship instead of a parasitic relationship. Some species are not able to reproduce in the absence of infection with Wolbachia. Thus, the use of existing strains of Wolbachia bacteria offers a potential strategy for the control of the population of mosquitoes and other pests and diseases. METHODS We searched ten databases and reviewed published papers regarding the role of Wolbachia as a promising drug target and emerging biological control agents of parasitic diseases between 1996 and 2017 (22 years) were considered eligible. Also, in the current study several patents (WO008652), (US7723062), and (US 0345249 A1) were reviewed. RESULTS Endosymbiotic Wolbachia bacteria, which are inherited from mothers, is transmitted to mosquitoes and interferes with pathogen transmission. They can change the reproduction of their host. Wolbachia is transmitted through the cytoplasm of eggs and have evolved different mechanisms for manipulating the reproduction of its hosts, including the induction of reproductive incompatibility, parthenogenesis, and feminization. The extensive effects of Wolbachia on reproduction and host fitness have made Wolbachia the issue of growing attention as a potential biocontrol agent. CONCLUSION Wolbachia has opened a new window to design a costly, potent and ecofriendly drug target for effective treatment and elimination of vector-borne parasitic diseases.
Collapse
Affiliation(s)
- Tooran Nayeri Chegeni
- Department of Parasitology, Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahdi Fakhar
- Department of Parasitology, Toxoplasmosis Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
35
|
Espinoza-Culupú A, Mendes E, Vitorino HA, da Silva PI, Borges MM. Mygalin: An Acylpolyamine With Bactericidal Activity. Front Microbiol 2020; 10:2928. [PMID: 31998255 PMCID: PMC6965172 DOI: 10.3389/fmicb.2019.02928] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/05/2019] [Indexed: 01/15/2023] Open
Abstract
Inappropriate use of antibiotics favors the selection and spread of resistant bacteria. To reduce the spread of these bacteria, finding new molecules with activity is urgent and necessary. Several polyamine analogs have been constructed and used to control microorganisms and tumor cells. Mygalin is a synthetic acylpolyamine, which are analogs of spermidine, derived from the hemolymph of the spider Acanthoscurria gomesiana. The effective activity of polyamines and their analogs has been associated with their structure. The presence of two acyl groups in the Mygalin structure may give this molecule a specific antibacterial activity. The aim of this study was to identify the mechanisms involved in the interaction of Mygalin with Escherichia coli to clarify its antimicrobial action. The results indicated that Mygalin exhibits intense dose and time-dependent bactericidal activity. Treatment of E. coli with this molecule caused membrane rupture, inhibition of DNA synthesis, DNA damage, and morphological changes. The esterase activity increased along with the intracellular production of reactive oxygen species (ROS) after treatment of the bacteria with Mygalin. In addition, this molecule was able to sequester iron and bind to LPS. We have shown that Mygalin has bactericidal activity with underlying mechanisms involving ROS generation and chelation of iron ions that are necessary for bacterial metabolism, which may contribute to its microbicidal activity. Taken together, our data suggest that Mygalin can be explored as a new alternative drug with antimicrobial potential against Gram-negative bacteria or other infectious agents.
Collapse
Affiliation(s)
- Abraham Espinoza-Culupú
- Ph.D. Program in Biotechnology, University of São Paulo, São Paulo, Brazil.,Bacteriology Laboratory, Butantan Institute, São Paulo, Brazil
| | | | - Hector Aguilar Vitorino
- Institute of Marine and Environmental Technology, University of Maryland Center for Environmental Science, Columbus Center, Baltimore, MD, United States
| | | | | |
Collapse
|
36
|
Vidović N, Recca T, Francescato P, Rabuffetti M, Sironi M, Oliva F, Pieraccini S, Speranza G. Conformational Studies on Two FtsZ Targeting Cyclic Peptides. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09962-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
37
|
Kusuma KD, Payne M, Ung AT, Bottomley AL, Harry EJ. FtsZ as an Antibacterial Target: Status and Guidelines for Progressing This Avenue. ACS Infect Dis 2019; 5:1279-1294. [PMID: 31268666 DOI: 10.1021/acsinfecdis.9b00055] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The disturbing increase in the number of bacterial pathogens that are resistant to multiple, or sometimes all, current antibiotics highlights the desperate need to pursue the discovery and development of novel classes of antibacterials. The wealth of knowledge available about the bacterial cell division machinery has aided target-driven approaches to identify new inhibitor compounds. The main division target being pursued is the highly conserved and essential protein FtsZ. Despite very active research on FtsZ inhibitors for several years, this protein is not yet targeted by any commercial antibiotic. Here, we discuss the suitability of FtsZ as an antibacterial target for drug development and review progress achieved in this area. We use hindsight to highlight the gaps that have slowed progress in FtsZ inhibitor development and to suggest guidelines for concluding that FtsZ is actually the target of these molecules, a key missing link in several studies. In moving forward, a multidisciplinary, communicative, and collaborative process, with sharing of research expertise, is critical if we are to succeed.
Collapse
|
38
|
FtsZ inhibitors as a new genera of antibacterial agents. Bioorg Chem 2019; 91:103169. [PMID: 31398602 DOI: 10.1016/j.bioorg.2019.103169] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 07/28/2019] [Accepted: 07/29/2019] [Indexed: 11/21/2022]
Abstract
The continuous emergence and rapid spread of a multidrug-resistant strain of bacterial pathogens have demanded the discovery and development of new antibacterial agents. A highly conserved prokaryotic cell division protein FtsZ is considered as a promising target by inhibiting bacterial cytokinesis. Inhibition of FtsZ assembly restrains the cell-division complex known as divisome, which results in filamentation, leading to lysis of the cell. This review focuses on details relating to the structure, function, and influence of FtsZ in bacterial cytokinesis. It also summarizes on the recent perspective of the known natural and synthetic inhibitors directly acting on FtsZ protein, with prominent antibacterial activities. A series of benzamides, trisubstituted benzimidazoles, isoquinolene, guanine nucleotides, zantrins, carbonylpyridine, 4 and 5-Substituted 1-phenyl naphthalenes, sulindac, vanillin analogues were studied here and recognized as FtsZ inhibitors that act either by disturbing FtsZ polymerization and/or GTPase activity. Doxorubicin, from a U.S. FDA, approved drug library displayed strong interaction with FtsZ. Several of the molecules discussed, include the prodrugs of benzamide based compound PC190723 (TXA-709 and TXA707). These molecules have exhibited the most prominent antibacterial activity against several strains of Staphylococcus aureus with minimal toxicity and good pharmacokinetics properties. The evidence of research reports and patent documentations on FtsZ protein has disclosed distinct support in the field of antibacterial drug discovery. The pressing need and interest shall facilitate the discovery of novel clinical molecules targeting FtsZ in the upcoming days.
Collapse
|
39
|
Qiu Y, Zhou L, Hu Y, Bao Y. Discovery of promising FtsZ inhibitors by E-pharmacophore, 3D-QSAR, molecular docking study, and molecular dynamics simulation. J Recept Signal Transduct Res 2019; 39:154-166. [PMID: 31355691 DOI: 10.1080/10799893.2019.1638404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Asbtract Filamentous temperature-sensitive protein Z (FtsZ), playing a key role in bacterial cell division, is regarded as a promising target for the design of antimicrobial agent. This study is looking for potential high-efficiency FtsZ inhibitors. Ligand-based pharmacophore and E-pharmacophore, virtual screening and molecular docking were used to detect promising FtsZ inhibitors, and molecular dynamics simulation was used to study the stability of protein-ligand complexes in this paper. Sixty-three inhibitors from published literatures with pIC50 ranging from 2.483 to 5.678 were collected to develop ligand-based pharmacophore model. 4DXD bound with 9PC was selected to develop the E-pharmacophore model. The pharmacophore models validated by test set method and decoy set were employed for virtual screening to exclude inactive compounds against ZINC database. After molecular docking, ADME analysis, IFD docking and MM-GBSA, 8 hits were identified as potent FtsZ inhibitors. A 50 ns molecular dynamics simulation was implemented on the compounds to assess the stability between potent inhibitors and FtsZ. The results indicated that the candidate compounds had a high docking score and were strongly combined with FtsZ by forming hydrogen bonding interactions with key amino acid residues, and van der Waals forces and hydrophobic interactions had significant contribution to the stability of the binding. Molecular dynamics simulation results showed that the protein-ligand compounds performed well in both the stability and flexibility of the simulation process.
Collapse
Affiliation(s)
- Yaping Qiu
- a College of Chemical Engineering, Sichuan University , Chengdu , China
| | - Lu Zhou
- a College of Chemical Engineering, Sichuan University , Chengdu , China
| | - Yanqiu Hu
- a College of Chemical Engineering, Sichuan University , Chengdu , China
| | - Yinfeng Bao
- a College of Chemical Engineering, Sichuan University , Chengdu , China
| |
Collapse
|
40
|
Urquhart AS, Hu J, Chooi YH, Idnurm A. The fungal gene cluster for biosynthesis of the antibacterial agent viriditoxin. Fungal Biol Biotechnol 2019; 6:2. [PMID: 31304040 PMCID: PMC6600887 DOI: 10.1186/s40694-019-0072-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 06/12/2019] [Indexed: 11/20/2022] Open
Abstract
Background Viriditoxin is one of the ‘classical’ secondary metabolites produced by fungi and that has antibacterial and other activities; however, the mechanism of its biosynthesis has remained unknown. Results Here, a gene cluster (vdt) responsible for viriditoxin synthesis was identified, via a bioinformatics analysis of the genomes of Paecilomyces variotii and Aspergillus viridinutans that both are viriditoxin producers. The function of the eight-membered gene cluster of P. variotii was characterized by targeted gene disruptions, revealing the roles of each gene in the synthesis of this molecule and establishing its biosynthetic pathway, which includes a Baeyer–Villiger monooxygenase catalyzed reaction. Additionally, a predicted catalytically-inactive hydrolase was identified as being required for the stereoselective biosynthesis of (M)-viriditoxin. The subcellular localizations of two proteins (VdtA and VdtG) were determined by fusing these proteins to green fluorescent protein, to establish that at least two intracellular structures are involved in the compartmentalization of the synthesis steps of this metabolite. Conclusions The predicted pathway for the synthesis of viriditoxin was established by a combination of genomics, bioinformatics, gene disruption and chemical analysis processes. Hence, this work reveals the basis for the synthesis of an understudied class of fungal secondary metabolites and provides a new model species for understanding the synthesis of biaryl compounds with a chiral axis. Electronic supplementary material The online version of this article (10.1186/s40694-019-0072-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrew S Urquhart
- 1School of BioSciences, University of Melbourne, Melbourne, Australia
| | - Jinyu Hu
- 2School of Molecular Sciences, University of Western Australia, Perth, Australia
| | - Yit-Heng Chooi
- 2School of Molecular Sciences, University of Western Australia, Perth, Australia
| | - Alexander Idnurm
- 1School of BioSciences, University of Melbourne, Melbourne, Australia
| |
Collapse
|
41
|
Hu J, Li H, Chooi YH. Fungal Dirigent Protein Controls the Stereoselectivity of Multicopper Oxidase-Catalyzed Phenol Coupling in Viriditoxin Biosynthesis. J Am Chem Soc 2019; 141:8068-8072. [DOI: 10.1021/jacs.9b03354] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Jinyu Hu
- School of Molecular Sciences, University of Western Australia, Perth, Western Australia 6009, Australia
| | - Hang Li
- School of Molecular Sciences, University of Western Australia, Perth, Western Australia 6009, Australia
| | - Yit-Heng Chooi
- School of Molecular Sciences, University of Western Australia, Perth, Western Australia 6009, Australia
| |
Collapse
|
42
|
Belete TM. Novel targets to develop new antibacterial agents and novel alternatives to antibacterial agents. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.humic.2019.01.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
43
|
Rivera-Chávez J, Caesar L, Garcia-Salazar JJ, Raja HA, Cech NB, Pearce CJ, Oberlies NH. Mycopyranone: a 8,8'-binaphthopyranone with potent anti-MRSA activity from the fungus Phialemoniopsis sp. Tetrahedron Lett 2019; 60:594-597. [PMID: 31598014 PMCID: PMC6785197 DOI: 10.1016/j.tetlet.2019.01.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
A new 8,8'-binaphthopyranone (mycopyranone, 1) was isolated from a solid fermentation of Phialemoniopsis sp. (fungal strain MSX61662), and the structure was elucidated via analysis of the NMR and HRESIMS data. The axial chirality of 1 was determined to be M by ECD. The central chirality at C-4/C-4' was assigned through a modified Mosher's method, while the absolute configuration at C-3/C-3' was deduced based on analysis of the 3 J H-3-H-4 values and NOESY correlations. Compound 1 was evaluated for its antimicrobial properties against Staphylococcus aureus SA1199 and a clinically relevant methicillin-resistant S. aureus strain (MRSA USA300 LAC strain AH1263). Compound 1 inhibited the growth of both strains in a concentration dependent manner with IC50 values in the low μM range. Molecular docking indicated that compound 1 binds to the FtsZ (tubulin-like) protein in the same pocket as viriditoxin (2), suggesting that 1 targets bacterial cell division.
Collapse
Affiliation(s)
- José Rivera-Chávez
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina 27412, USA
| | - Lindsay Caesar
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina 27412, USA
| | - Juan J Garcia-Salazar
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina 27412, USA
| | - Huzefa A Raja
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina 27412, USA
| | - Nadja B Cech
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina 27412, USA
| | - Cedric J Pearce
- Mycosynthetix, Inc., Hillsborough, North Carolina, 27278, USA
| | - Nicholas H Oberlies
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina 27412, USA
| |
Collapse
|
44
|
How to Study Antimicrobial Activities of Plant Extracts: A Critical Point of View. SUSTAINABLE DEVELOPMENT AND BIODIVERSITY 2018. [DOI: 10.1007/978-3-319-67045-4_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
45
|
Noh TH, Sen L, Hong J, Lee JH, Moon HR, Jung JH. Antibacterial activities of viriditoxin congeners and synthetic analogues against fish pathogens. Bioorg Med Chem Lett 2017; 27:4970-4974. [PMID: 29037949 DOI: 10.1016/j.bmcl.2017.10.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/06/2017] [Accepted: 10/07/2017] [Indexed: 11/30/2022]
Abstract
Viriditoxin is a fungal secondary metabolite of the fungus Paecilomyces variotii derived from the inner tissues of the giant jellyfish Nemopilema nomurai. Viriditoxin exhibits antibacterial activity against Streptococcus iniae and Streptococcus parauberis, which are major pathogens of aqua cultured fish. Viriditoxin induced abnormal cell morphologies in the fish pathogens S. iniae and S. parauberis, presumably by inhibiting FtsZ polymerization as was previously observed in Escherichia coli. Synthetic analogues of viriditoxin, designed based on docking simulation results to FtsZ of Staphylococcus aureus, were prepared and compared with viriditoxin for antibacterial activity. Reconstitution of free hydroxyl or carboxyl groups of the methoxyl or methyl ester groups of viriditoxin led to significant reduction of antibacterial activity, implying that the natural molecule is optimized for antibacterial activity to deter bacteria potentially harmful to Paecilomyces.
Collapse
Affiliation(s)
- Tae Hwan Noh
- College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Liu Sen
- College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Jongki Hong
- College of Pharmacy, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Joon-Hee Lee
- College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Hyung Ryong Moon
- College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Jee H Jung
- College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea.
| |
Collapse
|
46
|
Liu F, Venter H, Bi F, Semple SJ, Liu J, Jin C, Ma S. Synthesis and antibacterial activity of 5-methylphenanthridium derivatives as FtsZ inhibitors. Bioorg Med Chem Lett 2017. [DOI: 10.1016/j.bmcl.2017.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
47
|
Mathew B, Hobrath JV, Ross L, Connelly MC, Lofton H, Rajagopalan M, Guy RK, Reynolds RC. Screening and Development of New Inhibitors of FtsZ from M. Tuberculosis. PLoS One 2016; 11:e0164100. [PMID: 27768711 PMCID: PMC5074515 DOI: 10.1371/journal.pone.0164100] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 09/20/2016] [Indexed: 12/14/2022] Open
Abstract
A variety of commercial analogs and a newer series of Sulindac derivatives were screened for inhibition of M. tuberculosis (Mtb) in vitro and specifically as inhibitors of the essential mycobacterial tubulin homolog, FtsZ. Due to the ease of preparing diverse analogs and a favorable in vivo pharmacokinetic and toxicity profile of a representative analog, the Sulindac scaffold may be useful for further development against Mtb with respect to in vitro bacterial growth inhibition and selective activity for Mtb FtsZ versus mammalian tubulin. Further discovery efforts will require separating reported mammalian cell activity from both antibacterial activity and inhibition of Mtb FtsZ. Modeling studies suggest that these analogs bind in a specific region of the Mtb FtsZ polymer that differs from human tubulin and, in combination with a pharmacophore model presented herein, future hybrid analogs of the reported active molecules that more efficiently bind in this pocket may improve antibacterial activity while improving other drug characteristics.
Collapse
Affiliation(s)
- Bini Mathew
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, Alabama, 35205, United States of America
| | - Judith Varady Hobrath
- Drug Discovery Unit, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - Larry Ross
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, Alabama, 35205, United States of America
| | - Michele C. Connelly
- Dept. Chemical Biology & Therapeutics, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, United States of America
| | - Hava Lofton
- The University of Texas Health Science Center at Tyler, Tyler, Texas, 75708, United States of America
| | - Malini Rajagopalan
- The University of Texas Health Science Center at Tyler, Tyler, Texas, 75708, United States of America
| | - R. Kiplin Guy
- Dept. Chemical Biology & Therapeutics, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, United States of America
| | - Robert C. Reynolds
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
- Division of Hematology and Oncology, The University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
- * E-mail:
| |
Collapse
|
48
|
Broughton CE, Van Den Berg HA, Wemyss AM, Roper DI, Rodger A. Beyond the Discovery Void: New targets for antibacterial compounds. Sci Prog 2016; 99:153-182. [PMID: 28742471 PMCID: PMC10365418 DOI: 10.3184/003685016x14616130512308] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Antibiotics save many lives, but their efficacy is under threat: overprescription, population growth, and global travel all contribute to the rapid origination and spread of resistant strains. Exacerbating this threat is the fact that no new major classes of antibiotics have been discovered in the last 30 years: this is the "discovery void." We discuss the traditional molecular targets of antibiotics as well as putative novel targets.
Collapse
Affiliation(s)
| | | | - Alan M. Wemyss
- Molecular Organisation and Assembly in Cells Doctoral Training Centre
| | | | | |
Collapse
|
49
|
Hurley KA, Santos TMA, Nepomuceno GM, Huynh V, Shaw JT, Weibel DB. Targeting the Bacterial Division Protein FtsZ. J Med Chem 2016; 59:6975-98. [DOI: 10.1021/acs.jmedchem.5b01098] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Katherine A. Hurley
- Department of Pharmaceutical Sciences, University of Wisconsin—Madison, 777 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Thiago M. A. Santos
- Department
of Biochemistry, University of Wisconsin—Madison, 440 Henry Mall, Madison, Wisconsin 53706, United States
| | - Gabriella M. Nepomuceno
- Department of Chemistry, University of California—Davis, One Shields Avenue, Davis, California 95616, United States
| | - Valerie Huynh
- Department of Chemistry, University of California—Davis, One Shields Avenue, Davis, California 95616, United States
| | - Jared T. Shaw
- Department of Chemistry, University of California—Davis, One Shields Avenue, Davis, California 95616, United States
| | - Douglas B. Weibel
- Department
of Biochemistry, University of Wisconsin—Madison, 440 Henry Mall, Madison, Wisconsin 53706, United States
- Department of Chemistry, University of Wisconsin—Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
- Department of Biomedical Engineering, University of Wisconsin—Madison, 1550 Engineering Drive, Madison, Wisconsin 53706, United States
| |
Collapse
|
50
|
Liu Y, Kurtán T, Yun Wang C, Han Lin W, Orfali R, Müller WE, Daletos G, Proksch P. Cladosporinone, a new viriditoxin derivative from the hypersaline lake derived fungus Cladosporium cladosporioides. J Antibiot (Tokyo) 2016; 69:702-6. [PMID: 26905758 DOI: 10.1038/ja.2016.11] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/14/2016] [Accepted: 01/20/2016] [Indexed: 02/05/2023]
Abstract
A new cytotoxic viriditoxin derivative, cladosporinone (1), along with the known viriditoxin (2) and two viriditoxin derivatives (3 and 4) were obtained from the fungus Cladosporium cladosporioides isolated from the sediment of a hypersaline lake in Egypt. The structure of the new compound (1) was determined by 1D and 2D NMR measurements as well as by high-resolution ESIMS and electronic circular dichroism spectroscopy. All isolated compounds were studied for their cytotoxicity against the murine lymphoma cell line L5187Y and for their antibiotic activity against several pathogenic bacteria. Viriditoxin (2) was the most active compound in both bioassays. Compound 1 also exhibited strong cytotoxicity against the murine lymphoma cell line L5187Y with an IC50 value of 0.88 μm, whereas its antibiotic activity was weak.
Collapse
Affiliation(s)
- Yang Liu
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University, Duesseldorf, Germany.,Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, People's Republic of China
| | - Tibor Kurtán
- Department of Organic Chemistry, University of Debrecen, Debrecen, Hungary
| | - Chang Yun Wang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, People's Republic of China
| | - Wen Han Lin
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, People's Republic of China
| | - Raha Orfali
- Department of Pharmacognosy, Faculty of Pharmacy, King Saud University, Riyadh, KSA
| | - Werner Eg Müller
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Georgios Daletos
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University, Duesseldorf, Germany
| | - Peter Proksch
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University, Duesseldorf, Germany
| |
Collapse
|